77 results on '"Essalmani, R."'
Search Results
2. Schwann cells as a possible player in prion propagation
- Author
-
Archer, Fabienne, PERROT, GAETAN, Vilette, Didier, Le Dur, Annick, Besnard, N., Essalmani, R., Vilotte, Jean Luc, Paulin, D., BACHELIN, C., Baron, A., Laude, Hubert, Unité de recherche Génétique Biochimique et Cytogénétique (LGBC), and Institut National de la Recherche Agronomique (INRA)
- Subjects
[SDV]Life Sciences [q-bio] ,PRION ,SCHWANN CELLS ,CELLULES DE SCHWANN - Published
- 2002
3. Développement de nouveaux systèmes permissifs à l'agent de la tremblante ovine à partir de souris transgéniques
- Author
-
Archer, Fabienne, PERROT, GAETAN, Vilette, Didier, Le Dur, Annick, MADELAINE, M.F., COSTA DA SILVA, J., Soulier, S., Besnard, N., Essalmani, R., Vilotte, Jean Luc, Paulin, D., Laude, Hubert, Unité de recherche Virologie et Immunologie Moléculaires (VIM (UR 0892)), Institut National de la Recherche Agronomique (INRA), and Unité de recherche Génétique Biochimique et Cytogénétique (LGBC)
- Subjects
[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS - Abstract
National audience
- Published
- 2000
4. Développement de souris transgéniques exprimant l'allèle PRP-VRQ ovin, la protéine PRP murine ou bovine comme modèles d'étude in vivo de l'agent infectieux de la scrapie et autres ESST
- Author
-
Essalmani, R., Vilotte, Jean Luc, Soulier, S., Stinnakre, M.G., Lepourry, Laurence, COSTA DA SILVA, J., Besnard, N., Vaiman, Daniel, Petit, S., Rakotobe, Sabine, MADELAINE, M.F., Le Dur, Annick, Archer, A., Vilette, Didier, Laude, Didier, Unité de recherche Génétique Biochimique et Cytogénétique (LGBC), Institut National de la Recherche Agronomique (INRA), and Unité de recherche Virologie et Immunologie Moléculaires (VIM (UR 0892))
- Subjects
[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS - Abstract
National audience
- Published
- 2000
5. Transmission expérimentale et caractérisation des souches de tremblante naturelle: que peuvent apporter les souris transgéniques ?
- Author
-
Laude, Hubert, Le Dur, Annick, Rakotobe, Sabine, MADELAINE, M.F., Vilette, Didier, Besnard, N., Stinnakre, M.G., Essalmani, R., Soulier, S., Vilotte, Jean Luc, Unité de recherche Virologie et Immunologie Moléculaires (VIM (UR 0892)), Institut National de la Recherche Agronomique (INRA), and Unité de recherche Génétique Biochimique et Cytogénétique (LGBC)
- Subjects
[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS - Abstract
National audience
- Published
- 2000
6. Données de base sur la transgenèse
- Author
-
Essalmani, R., Soulier, S., Besnard, N., Hudrisier, Marthe, COSTA DA SILVA, J., Vilotte, Jean Luc, Unité de recherche Génétique Biochimique et Cytogénétique (LGBC), and Institut National de la Recherche Agronomique (INRA)
- Subjects
[SDV.SA]Life Sciences [q-bio]/Agricultural sciences ,BIOLOGIE MOLECULAIRE - Abstract
National audience; La transgenèse permet d’introduire dans le génome d’un animal un fragment d’ADN qui sera ensuite transmis de génération en génération. Elle utilise différentes approches méthodologiques. Certaines, encore limitées à peu d’espèces, permettent des modifications très fines du génome. La transgenèse, associée au développement de la biologie moléculaire, offre des applications multiples tant dans la recherche fondamentale qu’appliquée. Elle est de fait devenue un outil indispensable pour l’analyse de la régulation de l’expression des gènes et la compréhension de leur fonction.
- Published
- 2000
7. L'expression de l'allèle VRQ de la protéine ovine PrP confère une forte susceptibilité à l'agent de la tremblante du mouton à des souris transgéniques. Applications à d'autres protéines PrP et à l'obtention de lignées cellulaires
- Author
-
Vilotte, Jean Luc, Soulier, S., Essalmani, R., Stinnakre, M.G., Lepourry, Laurence, COSTA DA SILVA, J., Besnard, N., Vaiman, Daniel, Petit, S., Rakotobe, Sabine, Archer, Fabienne, MADELAINE, M.F., Le Dur, Annick, Vilette, Didier, Laude, Hubert, Unité de recherche Génétique Biochimique et Cytogénétique (LGBC), Institut National de la Recherche Agronomique (INRA), and Unité de recherche Virologie et Immunologie Moléculaires (VIM (UR 0892))
- Subjects
[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS - Abstract
National audience
- Published
- 2000
8. Transmission expérimentale de la tremblante du mouton : abolition de la barrière d'espèce chez des souris transgéniques exprimant le gène PRNP ovin
- Author
-
Vilotte, Jean Luc, Soulier, S., Stinnakre, M.G., Vaiman, Daniel, Lepourry, Laurence, COSTA DA SILVA, J., Besnard, N., Essalmani, R., Rakotobe, Sabine, Petit, S., MADELAINE, M.F., Le Dur, Annick, Vilette, Didier, Laude, Hubert, Unité de recherche Génétique Biochimique et Cytogénétique (LGBC), Institut National de la Recherche Agronomique (INRA), and Unité de recherche Virologie et Immunologie Moléculaires (VIM (UR 0892))
- Subjects
[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS - Abstract
National audience
- Published
- 2000
9. Transmission expérimentale de la tremblante du mouton à des souris transgéniques ovinisées : franchissement de la barrière d'espèce et mise au point d'outils de caractérisation de l'agent infectieux
- Author
-
Vilotte, Jean Luc, Soulier, S., Essalmani, R., Stinnakre, M.G., Lepourry, Laurence, COSTA DA SILVA, J., Besnard, N., Vaiman, Daniel, Petit, S., Rakotobe, Sabine, MADELAINE, M.F., Le Dur, Annick, Vilette, Didier, Laude, Hubert, Unité de recherche Génétique Biochimique et Cytogénétique (LGBC), Institut National de la Recherche Agronomique (INRA), and Unité de recherche Virologie et Immunologie Moléculaires (VIM (UR 0892))
- Subjects
[SDV]Life Sciences [q-bio] - Published
- 2000
10. Transgenic mice expressing ovine PrP are highly susceptible to scrapie
- Author
-
Vilotte, Jean Luc, Soulier, S., Stinnakre, M.G., Vaiman, Daniel, Lepourry, Laurence, COSTA DA SILVA, J., Besnard, N., Essalmani, R., Dawson, M., Buschmann, A., Groschup, Martin Hermann, Petit, S., MADELAINE, M.F., Vilette, Didier, Laude, Hubert, Unité de recherche Génétique Biochimique et Cytogénétique (LGBC), Institut National de la Recherche Agronomique (INRA), and Unité de recherche Virologie et Immunologie Moléculaires (VIM (UR 0892))
- Subjects
[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS - Abstract
International audience
- Published
- 1999
11. The activation and physiological functions of the proprotein convertases
- Author
-
SEIDAH, N, primary, MAYER, G, additional, ZAID, A, additional, ROUSSELET, E, additional, NASSOURY, N, additional, POIRIER, S, additional, ESSALMANI, R, additional, and PRAT, A, additional
- Published
- 2008
- Full Text
- View/download PDF
12. The role of presenilin-1 in the gamma-secretase cleavage of the amyloid precursor protein of Alzheimer's disease.
- Author
-
UCL - MD/FSIO - Département de physiologie et pharmacologie, Octave, Jean-Noël, Essalmani, R, Tasiaux, Bernadette, Menager, J, Czech, C., Mercken, L., UCL - MD/FSIO - Département de physiologie et pharmacologie, Octave, Jean-Noël, Essalmani, R, Tasiaux, Bernadette, Menager, J, Czech, C., and Mercken, L.
- Abstract
Presenilin-1 (PS1) is required for the release of the intracellular domain of Notch from the plasma membrane as well as for the cleavage of the amyloid precursor protein (APP) at the gamma-secretase cleavage site. It remains to be demonstrated whether PS1 acts by facilitating the activity of the protease concerned or is the protease itself. PS1 could have a gamma-secretase activity by itself or could traffic APP and Notch to the appropriate cellular compartment for processing. Human APP 695 and PS1 were coexpressed in Sf9 insect cells, in which endogenous gamma-secretase activity is not detected. In baculovirus-infected Sf9 cells, PS1 undergoes endoproteolysis and interacts with APP. However, PS1 does not cleave APP in Sf9 cells. In CHO cells, endocytosis of APP is required for Abeta secretion. Deletion of the cytoplasmic sequence of APP (APPDeltaC) inhibits both APP endocytosis and Abeta production. When APPDeltaC and PS1 are coexpressed in CHO cells, Abeta is secreted without endocytosis of APP. Taken together, these results conclusively show that, although PS1 does not cleave APP in Sf9 cells, PS1 allows the secretion of Abeta without endocytosis of APP by CHO cells.
- Published
- 2000
13. A Bovine Prion Acquires an Epidemic Bovine Spongiform Encephalopathy Strain-Like Phenotype on Interspecies Transmission
- Author
-
Beringue, V., primary, Andreoletti, O., additional, Le Dur, A., additional, Essalmani, R., additional, Vilotte, J.-L., additional, Lacroux, C., additional, Reine, F., additional, Herzog, L., additional, Biacabe, A.-G., additional, Baron, T., additional, Caramelli, M., additional, Casalone, C., additional, and Laude, H., additional
- Published
- 2007
- Full Text
- View/download PDF
14. The long term adenoviral expression of the human amyloid precursor protein shows different secretase activities in rat cortical neurons and astrocytes.
- Author
-
UCL - MD/FSIO - Département de physiologie et pharmacologie, Macq, A F, Czech, C., Essalmani, R, Brion, J P, Maron, A, Mercken, L., Pradier, L., Octave, Jean-Noël, UCL - MD/FSIO - Département de physiologie et pharmacologie, Macq, A F, Czech, C., Essalmani, R, Brion, J P, Maron, A, Mercken, L., Pradier, L., and Octave, Jean-Noël
- Abstract
Recombinant adenoviruses were used for the expression of human amyloid precursor protein (APP) of Alzheimer's disease in primary cultures of rat cortical neurons and astrocytes. The catabolic pathways of human APP were studied 3 to 4 days after infection, when the equilibrium of APP production was reached. Although the expression of human wild type APP (WtAPP) by rat neurons induced the production of both extracellular and intraneuronal amyloid peptide (Abeta), Abeta was not detected in the culture medium of rat astrocytes producing human WtAPP. Because a low beta-secretase activity was previously reported in rodent astrocytes, we wondered whether modifications of the APP amino acid sequence at the beta-secretase clipping site would modify the astrocytic production of Abeta. Interestingly, rat astrocytes produced high amounts of Abeta after expression of human APP carrying a double amino acid substitution responsible for Alzheimer's disease in a large Swedish family (SwAPP). In both rat cortical neurons and astrocytes, the beta-secretase cleavage of the human SwAPP occurred very early in the secretion process in a cellular compartment in which a different sorting of SwAPP and WtAPP seems unlikely. These results suggest that human WtAPP and SwAPP could be processed by different beta-secretase activities.
- Published
- 1998
15. The long term adenoviral expression of the human amyloid precursor protein shows different secretase activities in rat cortical neurons and astrocytes.
- Author
-
Macq, Anne Francoise, Czech, C, Essalmani, R, Brion, Jean Pierre, Maron, A, Mercken, Luc, Pradier, L, Octave, Jean Noël, Macq, Anne Francoise, Czech, C, Essalmani, R, Brion, Jean Pierre, Maron, A, Mercken, Luc, Pradier, L, and Octave, Jean Noël
- Abstract
Recombinant adenoviruses were used for the expression of human amyloid precursor protein (APP) of Alzheimer's disease in primary cultures of rat cortical neurons and astrocytes. The catabolic pathways of human APP were studied 3 to 4 days after infection, when the equilibrium of APP production was reached. Although the expression of human wild type APP (WtAPP) by rat neurons induced the production of both extracellular and intraneuronal amyloid peptide (Abeta), Abeta was not detected in the culture medium of rat astrocytes producing human WtAPP. Because a low beta-secretase activity was previously reported in rodent astrocytes, we wondered whether modifications of the APP amino acid sequence at the beta-secretase clipping site would modify the astrocytic production of Abeta. Interestingly, rat astrocytes produced high amounts of Abeta after expression of human APP carrying a double amino acid substitution responsible for Alzheimer's disease in a large Swedish family (SwAPP). In both rat cortical neurons and astrocytes, the beta-secretase cleavage of the human SwAPP occurred very early in the secretion process in a cellular compartment in which a different sorting of SwAPP and WtAPP seems unlikely. These results suggest that human WtAPP and SwAPP could be processed by different beta-secretase activities., Journal Article, Research Support, Non-U.S. Gov't, info:eu-repo/semantics/published
- Published
- 1998
16. Baculovirus-infected cells do not produce the amyloid peptide of Alzheimer's disease from its precursor.
- Author
-
UCL - MD/FSIO - Département de physiologie et pharmacologie, Essalmani, R., Guillaume, J. M., Mercken, L., Octave, Jean-Noël, UCL - MD/FSIO - Département de physiologie et pharmacologie, Essalmani, R., Guillaume, J. M., Mercken, L., and Octave, Jean-Noël
- Abstract
The amyloid peptide (Abeta) of Alzheimer's disease (AD) is produced by proteolytic cleavage of a larger precursor, the amyloid peptide precursor or APP. The discovery of pathogenic mutations in the APP gene provides strong evidence for the hypothesis that APP metabolism is involved in the etiology of AD. To study the metabolism of the protein, human APP has been expressed in several mammalian cell types. Insect cells, infected by a recombinant baculovirus carrying the human APP sequence, also provide an interesting expression system because these cells do not produce endogenous APP. Baculovirus-infected cells synthesize very high amounts of extracellular soluble APP, after cleavage of the transmembrane protein, as described for mammalian cells. However, we demonstrate here that insect cells do not produce Abeta from APP. These results suggest that while the enzymatic activity needed for the production of soluble APP is conserved between insect and mammalian cells, the enzymes required for the production of Abeta from APP are only expressed in mammalian cells.
- Published
- 1996
17. Missense mutations associated with familial Alzheimer's disease in Sweden lead to the production of the amyloid peptide without internalization of its precursor.
- Author
-
UCL - MD/FSIO - Département de physiologie et pharmacologie, Essalmani, R., Macq, A. F., Mercken, L., Octave, Jean-Noël, UCL - MD/FSIO - Département de physiologie et pharmacologie, Essalmani, R., Macq, A. F., Mercken, L., and Octave, Jean-Noël
- Abstract
Production of soluble amyloid peptide precursor (APP) and amyloid peptide (A beta) was measured in CHO cells transfected by the wild-type APP 695 cDNA sequence or by the same sequence carrying missense mutations associated with familial Alzheimer's disease in Sweden. Deletion of the C-terminal domain of the protein corresponding to residues 654 to 695 of APP 695 not only inhibited very significantly the internalization of APP at 37 degrees C, but also led to the secretion of an uncleaved APP in the culture medium of CHO cells. This deletion did not affect A beta production from the Swedish APP but was able to inhibit the production of the wild-type APP. These results demonstrate that, in CHO cells, the internalization of the wild-type APP is needed for A beta production, while the production of the amyloid peptide from Swedish APP is independent of the internalization process.
- Published
- 1996
18. Données de base sur la transgenèse
- Author
-
ESSALMANI, R., primary, SOULIER, S., additional, BESNARD, N., additional, HUDRISIER, M., additional, COSTA DA SILVA, J., additional, and VILOTTE, J.L., additional
- Published
- 2000
- Full Text
- View/download PDF
19. Baculovirus‐infected cells do not produce the amyloid peptide of Alzheimer's disease from its precursor
- Author
-
Essalmani, R., primary, Guillaume, J.M., additional, Mercken, L., additional, and Octave, J.N., additional
- Published
- 1996
- Full Text
- View/download PDF
20. The role of presenilin-1 in the gamma-secretase cleavage of the amyloid precursor protein of Alzheimer's disease.
- Author
-
Octave, J N, Essalmani, R, Tasiaux, B, Menager, J, Czech, C, and Mercken, L
- Abstract
Presenilin-1 (PS1) is required for the release of the intracellular domain of Notch from the plasma membrane as well as for the cleavage of the amyloid precursor protein (APP) at the gamma-secretase cleavage site. It remains to be demonstrated whether PS1 acts by facilitating the activity of the protease concerned or is the protease itself. PS1 could have a gamma-secretase activity by itself or could traffic APP and Notch to the appropriate cellular compartment for processing. Human APP 695 and PS1 were coexpressed in Sf9 insect cells, in which endogenous gamma-secretase activity is not detected. In baculovirus-infected Sf9 cells, PS1 undergoes endoproteolysis and interacts with APP. However, PS1 does not cleave APP in Sf9 cells. In CHO cells, endocytosis of APP is required for Abeta secretion. Deletion of the cytoplasmic sequence of APP (APPDeltaC) inhibits both APP endocytosis and Abeta production. When APPDeltaC and PS1 are coexpressed in CHO cells, Abeta is secreted without endocytosis of APP. Taken together, these results conclusively show that, although PS1 does not cleave APP in Sf9 cells, PS1 allows the secretion of Abeta without endocytosis of APP by CHO cells.
- Published
- 2000
21. The long term adenoviral expression of the human amyloid precursor protein shows different secretase activities in rat cortical neurons and astrocytes.
- Author
-
Macq, A F, Czech, C, Essalmani, R, Brion, J P, Maron, A, Mercken, L, Pradier, L, and Octave, J N
- Abstract
Recombinant adenoviruses were used for the expression of human amyloid precursor protein (APP) of Alzheimer's disease in primary cultures of rat cortical neurons and astrocytes. The catabolic pathways of human APP were studied 3 to 4 days after infection, when the equilibrium of APP production was reached. Although the expression of human wild type APP (WtAPP) by rat neurons induced the production of both extracellular and intraneuronal amyloid peptide (Abeta), Abeta was not detected in the culture medium of rat astrocytes producing human WtAPP. Because a low beta-secretase activity was previously reported in rodent astrocytes, we wondered whether modifications of the APP amino acid sequence at the beta-secretase clipping site would modify the astrocytic production of Abeta. Interestingly, rat astrocytes produced high amounts of Abeta after expression of human APP carrying a double amino acid substitution responsible for Alzheimer's disease in a large Swedish family (SwAPP). In both rat cortical neurons and astrocytes, the beta-secretase cleavage of the human SwAPP occurred very early in the secretion process in a cellular compartment in which a different sorting of SwAPP and WtAPP seems unlikely. These results suggest that human WtAPP and SwAPP could be processed by different beta-secretase activities.
- Published
- 1998
22. Corrigendum to "PCSK7: A novel regulator of apolipoprotein B and a potential target against non-alcoholic fatty liver disease" [Metabolism Volume 150, January 2024, 155736, PMID: 7967646].
- Author
-
Sachan V, LeDévéhat M, Roubtsova A, Essalmani R, Laurendeau JF, Garçon D, Susan-Sesiga D, Duval S, Mikaeeli S, Hamelin J, Evagelidis A, Chong M, Paré G, Chernetsova E, Gao ZH, Robillard I, Ruiz M, Trinh VQ, Estall JL, Faraj M, Austin RC, Sauvageau M, Prat A, Kiss RS, and Seidah NG
- Published
- 2024
- Full Text
- View/download PDF
23. Insights into PCSK9-LDLR Regulation and Trafficking via the Differential Functions of MHC-I Proteins HFE and HLA-C.
- Author
-
Mikaeeli S, Ben Djoudi Ouadda A, Evagelidis A, Essalmani R, Ramos OHP, Fruchart-Gaillard C, and Seidah NG
- Subjects
- Humans, HEK293 Cells, Protein Binding, Receptors, LDL metabolism, Proprotein Convertase 9 metabolism, Proprotein Convertase 9 genetics, Protein Transport, Hemochromatosis Protein metabolism, Hemochromatosis Protein genetics, HLA-C Antigens metabolism, Lysosomes metabolism
- Abstract
PCSK9 is implicated in familial hypercholesterolemia via targeting the cell surface PCSK9-LDLR complex toward lysosomal degradation. The M2 repeat in the PCSK9's C-terminal domain is essential for its extracellular function, potentially through its interaction with an unidentified "protein X". The M2 repeat was recently shown to bind an R-x-E motif in MHC-class-I proteins (implicated in the immune system), like HLA-C, and causing their lysosomal degradation. These findings suggested a new role of PCSK9 in the immune system and that HLA-like proteins could be "protein X" candidates. However, the participation of each member of the MHC-I protein family in this process and their regulation of PCSK9's function have yet to be determined. Herein, we compared the implication of MHC-I-like proteins such as HFE (involved in iron homeostasis) and HLA-C on the extracellular function of PCSK9. Our data revealed that the M2 domain regulates the intracellular sorting of the PCSK9-LDLR complex to lysosomes, and that HFE is a new target of PCSK9 that inhibits its activity on the LDLR, whereas HLA-C enhances its function. This work suggests the potential modulation of PCSK9's functions through interactions of HFE and HLA-C.
- Published
- 2024
- Full Text
- View/download PDF
24. PCSK7: A novel regulator of apolipoprotein B and a potential target against non-alcoholic fatty liver disease.
- Author
-
Sachan V, Le Dévéhat M, Roubtsova A, Essalmani R, Laurendeau JF, Garçon D, Susan-Resiga D, Duval S, Mikaeeli S, Hamelin J, Evagelidis A, Chong M, Paré G, Chernetsova E, Gao ZH, Robillard I, Ruiz M, Trinh VQ, Estall JL, Faraj M, Austin RC, Sauvageau M, Prat A, Kiss RS, and Seidah NG
- Subjects
- Mice, Animals, Subtilisin metabolism, Triglycerides metabolism, Liver metabolism, Apolipoproteins B genetics, Apolipoproteins B metabolism, Proprotein Convertases metabolism, Apolipoprotein B-100 genetics, Apolipoprotein B-100 metabolism, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Background: Epidemiological evidence links the proprotein convertase subtilisin/kexin 7 (PCSK7) to triglyceride (TG) metabolism. We associated the known PCSK7 gain-of-function non-coding SNP rs236918 with higher levels of plasma apolipoprotein B (apoB) and the loss-of-function coding variant p.Pro777Leu (SNP rs201598301) with lower apoB and TG. Herein, we aimed to unravel the in vivo role of liver PCSK7., Methods: We biochemically defined the functional role of PCSK7 in lipid metabolism using hepatic cell lines and Pcsk7
-/- mice. Our findings were validated following subcutaneous administration of hepatocyte-targeted N-acetylgalactosamine (GalNAc)-antisense oligonucleotides (ASOs) against Pcsk7., Results: Independent of its proteolytic activity, membrane-bound PCSK7 binds apoB100 in the endoplasmic reticulum and enhances its secretion. Mechanistically, the loss of PCSK7/Pcsk7 leads to apoB100 degradation, triggering an unfolded protein response, autophagy, and β-oxidation, eventually reducing lipid accumulation in hepatocytes. Non-alcoholic fatty liver disease (NAFLD) was induced by a 12-week high fat/fructose/cholesterol diet in wild type (WT) and Pcsk7-/- mice that were then allowed to recover on a 4-week control diet. Pcsk7-/- mice recovered more effectively than WT mice from all NAFLD-related liver phenotypes. Finally, subcutaneous administration of GalNAc-ASOs targeting hepatic Pcsk7 to WT mice validated the above results., Conclusions: Our data reveal hepatic PCSK7 as one of the major regulators of apoB, and its absence reduces apoB secretion from hepatocytes favoring its ubiquitination and degradation by the proteasome. This results in a cascade of events, eventually reducing hepatic lipid accumulation, thus supporting the notion of silencing PCSK7 mRNA in hepatocytes for targeting NAFLD., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Nabil G Seidah has patent METHOD FOR REDUCING HEPATIC TRIGLYCERIDES pending to Nabil G Seidah., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
25. SKI-1/S1P Facilitates SARS-CoV-2 Spike Induced Cell-to-Cell Fusion via Activation of SREBP-2 and Metalloproteases, Whereas PCSK9 Enhances the Degradation of ACE2.
- Author
-
Essalmani R, Andréo U, Evagelidis A, Le Dévéhat M, Pereira Ramos OH, Fruchart Gaillard C, Susan-Resiga D, Cohen ÉA, and Seidah NG
- Subjects
- Humans, Angiotensin-Converting Enzyme 2, Cell Fusion, HeLa Cells, Metalloproteases, SARS-CoV-2, Sterol Regulatory Element Binding Protein 1, COVID-19, Proprotein Convertase 9 genetics
- Abstract
Proprotein convertases activate various envelope glycoproteins and participate in cellular entry of many viruses. We recently showed that the convertase furin is critical for the infectivity of SARS-CoV-2, which requires cleavage of its spike protein (S) at two sites: S1/S2 and S2'. This study investigates the implication of the two cholesterol-regulating convertases SKI-1 and PCSK9 in SARS-CoV-2 entry. The assays used were cell-to-cell fusion in HeLa cells and pseudoparticle entry into Calu-3 cells. SKI-1 increased cell-to-cell fusion by enhancing the activation of SREBP-2, whereas PCSK9 reduced cell-to-cell fusion by promoting the cellular degradation of ACE2. SKI-1 activity led to enhanced S2' formation, which was attributed to increased metalloprotease activity as a response to enhanced cholesterol levels via activated SREBP-2. However, high metalloprotease activity resulted in the shedding of S2' into a new C-terminal fragment (S2″), leading to reduced cell-to-cell fusion. Indeed, S-mutants that increase S2″ formation abolished S2' and cell-to-cell fusion, as well as pseudoparticle entry, indicating that the formation of S2″ prevents SARS-CoV-2 cell-to-cell fusion and entry. We next demonstrated that PCSK9 enhanced the cellular degradation of ACE2, thereby reducing cell-to-cell fusion. However, different from the LDLR, a canonical target of PCSK9, the C-terminal CHRD domain of PCSK9 is dispensable for the PCSK9-induced degradation of ACE2. Molecular modeling suggested the binding of ACE2 to the Pro/Catalytic domains of mature PCSK9. Thus, both cholesterol-regulating convertases SKI-1 and PCSK9 can modulate SARS-CoV-2 entry via two independent mechanisms.
- Published
- 2023
- Full Text
- View/download PDF
26. Erratum for Essalmani et al., "Distinctive Roles of Furin and TMPRSS2 in SARS-CoV-2 Infectivity".
- Author
-
Essalmani R, Jain J, Susan-Resiga D, Andréo U, Evagelidis A, Derbali RM, Huynh DN, Dallaire F, Laporte M, Delpal A, Sutto-Ortiz P, Coutard B, Mapa C, Wilcoxen K, Decroly E, Pham TNQ, Cohen ÉA, and Seidah NG
- Published
- 2022
- Full Text
- View/download PDF
27. Distinctive Roles of Furin and TMPRSS2 in SARS-CoV-2 Infectivity.
- Author
-
Essalmani R, Jain J, Susan-Resiga D, Andréo U, Evagelidis A, Derbali RM, Huynh DN, Dallaire F, Laporte M, Delpal A, Sutto-Ortiz P, Coutard B, Mapa C, Wilcoxen K, Decroly E, Nq Pham T, Cohen ÉA, and Seidah NG
- Subjects
- Angiotensin-Converting Enzyme 2 metabolism, Animals, HeLa Cells, Humans, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Virus Internalization, COVID-19 pathology, COVID-19 virology, Furin metabolism, SARS-CoV-2 genetics, SARS-CoV-2 pathogenicity, Serine Endopeptidases metabolism
- Abstract
The spike protein (S) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directs infection of the lungs and other tissues following its binding to the angiotensin-converting enzyme 2 (ACE2) receptor. For effective infection, the S protein is cleaved at two sites: S1/S2 and S2'. The "priming" of the surface S protein at S1/S2 (P R RA R
685 ↓) [the underlined basic amino acids refer to critical residues needed for the furin recognition] by furin has been shown to be important for SARS-CoV-2 infectivity in cells and small-animal models. In this study, for the first time we unambiguously identified by proteomics the fusion activation site S2' as KPS KR815 ↓ (the underlined basic amino acids refer to critical residues needed for the furin recognition) and demonstrated that this cleavage was strongly enhanced by ACE2 engagement with the S protein. Novel pharmacological furin inhibitors (BOS inhibitors) effectively blocked endogenous S protein processing at both sites in HeLa cells, and SARS-CoV-2 infection of lung-derived Calu-3 cells was completely prevented by combined inhibitors of furin (BOS) and type II transmembrane serine protease 2 (TMPRSS2) (camostat). Quantitative analyses of cell-to-cell fusion and S protein processing revealed that ACE2 shedding by TMPRSS2 was required for TMPRSS2-mediated enhancement of fusion in the absence of S1/S2 priming. We further demonstrated that the collectrin dimerization domain of ACE2 was essential for the effect of TMPRSS2 on cell-to-cell fusion. Overall, our results indicate that furin and TMPRSS2 act synergistically in viral entry and infectivity, supporting the combination of furin and TMPRSS2 inhibitors as potent antivirals against SARS-CoV-2. IMPORTANCE SARS-CoV-2, the etiological agent of COVID-19, has so far resulted in >6.1 million deaths worldwide. The spike protein (S) of the virus directs infection of the lungs and other tissues by binding the angiotensin-converting enzyme 2 (ACE2) receptor. For effective infection, the S protein is cleaved at two sites: S1/S2 and S2'. Cleavage at S1/S2 induces a conformational change favoring the S protein recognition by ACE2. The S2' cleavage is critical for triggering membrane fusion and virus entry into host cells. Our study highlights the complex dynamics of interaction between the S protein, ACE2, and the host proteases furin and TMPRSS2 during SARS-CoV-2 entry and suggests that the combination of a nontoxic furin inhibitor with a TMPRSS2 inhibitor significantly reduces viral entry in lung cells, as evidenced by an average synergistic ∼95% reduction of viral infection. This represents a powerful novel antiviral approach to reduce viral spread in individuals infected by SARS-CoV-2 or future related coronaviruses.- Published
- 2022
- Full Text
- View/download PDF
28. Asialoglycoprotein receptor 1 is a novel PCSK9-independent ligand of liver LDLR cleaved by furin.
- Author
-
Susan-Resiga D, Girard E, Essalmani R, Roubtsova A, Marcinkiewicz J, Derbali RM, Evagelidis A, Byun JH, Lebeau PF, Austin RC, and Seidah NG
- Subjects
- Animals, Asialoglycoprotein Receptor genetics, Furin genetics, HEK293 Cells, Hep G2 Cells, Humans, Mice, Mice, Knockout, Proprotein Convertase 9 genetics, Receptors, LDL genetics, Asialoglycoprotein Receptor metabolism, Furin metabolism, Liver metabolism, Proprotein Convertase 9 metabolism, Receptors, LDL metabolism
- Abstract
The hepatic carbohydrate-recognizing asialoglycoprotein receptor (ASGR1) mediates the endocytosis/lysosomal degradation of desialylated glycoproteins following binding to terminal galactose/N-acetylgalactosamine. Human heterozygote carriers of ASGR1 deletions exhibit ∼34% lower risk of coronary artery disease and ∼10% to 14% reduction of non-HDL cholesterol. Since the proprotein convertase PCSK9 is a major degrader of the low-density lipoprotein receptor (LDLR), we investigated the degradation and functionality of LDLR and/or PCSK9 by endogenous/overexpressed ASGR1 using Western blot and immunofluorescence in HepG2-naïve and HepG2-PCSK9-knockout cells. ASGR1, like PCSK9, targets LDLR, and both independently interact with/enhance the degradation of the receptor. This lack of cooperativity between PCSK9 and ASGR1 was confirmed in livers of wildtype (WT) and Pcsk9
-/- mice. ASGR1 knockdown in HepG2-naïve cells significantly increased total (∼1.2-fold) and cell-surface (∼4-fold) LDLR protein. In HepG2-PCSK9-knockout cells, ASGR1 silencing led to ∼2-fold higher levels of LDLR protein and DiI (1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate)-LDL uptake associated with ∼9-fold increased cell-surface LDLR. Overexpression of WT-ASGR1/2 primarily reduced levels of immature non-O-glycosylated LDLR (∼110 kDa), whereas the triple Ala-mutant of Gln240/Trp244/Glu253 (characterized by loss of carbohydrate binding) reduced expression of the mature form of LDLR (∼150 kDa), suggesting that ASGR1 binds the LDLR in both a sugar-dependent and -independent fashion. The protease furin cleaves ASGR1 at the RKMK103 ↓ motif into a secreted form, likely resulting in a loss of function on LDLR. Altogether, we demonstrate that LDLR is the first example of a liver-receptor ligand of ASGR1. We conclude that silencing of ASGR1 and PCSK9 may lead to higher LDL uptake by hepatocytes, thereby providing a novel approach to further reduce LDL cholesterol levels., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article. R. C. A. is a Career Investigator of the Heart and Stroke Foundation of Ontario and holds the Amgen Canada Research Chair in the Division of Nephrology at St. Joseph’s Healthcare and McMaster University., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
29. Substantial PCSK9 inactivation in β-cells does not modify glucose homeostasis or insulin secretion in mice.
- Author
-
Peyot ML, Roubtsova A, Lussier R, Chamberland A, Essalmani R, Murthy Madiraju SR, Seidah NG, Prentki M, and Prat A
- Subjects
- Animals, Male, Mice, Cholesterol metabolism, Insulin metabolism, Mice, Knockout, Glucose metabolism, Homeostasis, Insulin Secretion drug effects, Insulin-Secreting Cells metabolism, Insulin-Secreting Cells drug effects, Proprotein Convertase 9 genetics, Proprotein Convertase 9 metabolism, Receptors, LDL metabolism, Receptors, LDL genetics
- Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in cholesterol homeostasis by promoting the degradation of the LDL receptor (LDLR). PCSK9 loss-of-function mutations are associated with increased fasting plasma glucose levels and slightly elevated risk of type 2-diabetes. Considering the known detrimental effects of cholesterol accumulation in β-cell, and the widespread use of PCSK9 inhibitors to treat hypercholesterolemia, it is important to gain insight into the role of pancreatic PCSK9 in glucose homeostasis and β-cell function. We generated the first β-cell-specific KO of PCSK9 (βKO). PCSK9 mRNA and protein expression were reduced by 48% and 78% in βKO islets, respectively, indicating that β-cells constitute a major site of PCSK9 expression. In islets, loss of β-cell PCSK9 resulted in unchanged LDLR protein levels, but reduced LDLR mRNA, indicating that cholesterol internalization is enhanced and that β-cell PCSK9 promotes LDLR degradation. In contrast, whole body PCSK9 KO mice exhibited 2-fold higher LDLR protein levels in islets and a stable expression of cholesterogenic genes. Whole body KO and βKO mice presented normal glucose tolerance, insulin release in response to glucose load and insulin sensitivity. Ex vivo glucose-stimulated insulin secretion in presence or absence of fatty acids was similar in WT and KO islets. Like KO mice, individuals carrying loss-of-function PCSK9 variants may be protected from cholesterol-induced toxicity due to reduced circulating cholesterol levels. Using both whole body KO or βKO models, our data demonstrate that PCSK9 deletion in mouse does not have any toxic effect on β-cell function and glucose homeostasis., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. In Vivo Analysis of the Contribution of Proprotein Convertases to the Processing of FGF23.
- Author
-
Al Rifai O, Susan-Resiga D, Essalmani R, Creemers JWM, Seidah NG, and Ferron M
- Subjects
- Animals, Bone Marrow metabolism, Fibroblast Growth Factor-23 genetics, Furin genetics, Iron Deficiencies genetics, Iron Deficiencies metabolism, Kidney metabolism, Liver metabolism, Mice, Mice, Knockout, Proprotein Convertase 5 genetics, Fibroblast Growth Factor-23 metabolism, Furin metabolism, Osteoblasts metabolism, Osteocytes metabolism, Proprotein Convertase 5 metabolism
- Abstract
Fibroblast growth factor 23 (FGF23) is a hormone secreted from fully differentiated osteoblasts and osteocytes that inhibits phosphate reabsorption by kidney proximal tubules. The full-length (i.e., intact) protein mediates FGF23 endocrine functions, while endoproteolytic cleavage at a consensus cleavage sequence for the proprotein convertases (PCs) inactivates FGF23. Two PCs, furin and PC5, were shown to cleave FGF23 in vitro at RHTR
179 ↓, but whether they are fulfilling this function in vivo is currently unknown. To address this question, we used here mice lacking either or both furin and PC5 in cell-specific manners and mice lacking the paired basic amino acid-cleaving enzyme 4 (PACE4) in all cells. Our analysis shows that furin inactivation in osteoblasts and osteocytes results in a 25% increase in circulating intact FGF23, without any significant impact on serum phosphate levels, whether mice are maintained on a normal or a low phosphate diet. Under conditions of iron deficiency, FGF23 is normally processed in control mice, but its processing is impaired in mice lacking furin in osteoblasts and osteocytes. In contrast, FGF23 is normally cleaved following erythropoietin or IL-1β injections in mice lacking furin or both furin and PC5, and in PACE4-deficient mice. Altogether, these studies suggest that furin is only partially responsible for FGF23 cleavage under certain conditions in vivo . The processing of FGF23 may therefore involve the redundant action of multiple PCs or of other peptidases in osteoblasts, osteocytes and hematopoietic cells., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Al Rifai, Susan-Resiga, Essalmani, Creemers, Seidah and Ferron.)- Published
- 2021
- Full Text
- View/download PDF
31. Proprotein convertase 7 (PCSK7) reduces apoA-V levels.
- Author
-
Ashraf Y, Duval S, Sachan V, Essalmani R, Susan-Resiga D, Roubtsova A, Hamelin J, Gerhardy S, Kirchhofer D, Tagliabracci VS, Prat A, Kiss RS, and Seidah NG
- Subjects
- Animals, Apolipoprotein A-V blood, Cell Line, Tumor, Endoplasmic Reticulum metabolism, Hepatocytes metabolism, Humans, Lysosomes metabolism, Mice, Inbred C57BL, Mice, Knockout, Polymorphism, Single Nucleotide, Subtilisins metabolism, Triglycerides blood, Exome Sequencing methods, Apolipoprotein A-V metabolism, Liver metabolism, Subtilisins genetics, Triglycerides metabolism
- Abstract
The locus of the human proprotein convertase subtilisin-kexin type-7 (PC7) gene (PCSK7) is on chromosome 11q23.3 close to the gene cluster APOA5/APOA4/APOC3/APOA1, a region implicated in the regulation of lipoprotein metabolism. A GWAS reported the association of PCSK7 SNPs with plasma triglyceride (TG), and exome sequencing of African Americans revealed the association of a low-frequency coding variant of PC7 (R504H; SNP rs142953140) with a ~ 30% TG reduction. Another PCSK7 SNP rs508487 is in linkage disequilibrium with a promoter variant of the liver-derived apolipoprotein A-V (apoA-V), an indirect activator of the lipoprotein lipase (LpL), and is associated with elevated TG levels. We thus hypothesized that PC7 regulates the levels/activity of apoA-V. Studies in the human hepatic cell line HuH7 revealed that wild-type (WT) PC7 and its endoplasmic reticulum (ER)-retained forms bind to and enhance the degradation of human apoA-V in acidic lysosomes in a nonenzymatic fashion. PC7-induced degradation of apoA-V is inhibited by bafilomycin A1 and the alkalinizing agents: chloroquine and NH
4 Cl. Thus, the PC7-induced apoA-V degradation implicates an ER-lysosomal communication inhibited by bafilomycin A1. In vitro, the natural R504H mutant enhances PC7 Ser505 phosphorylation at the structurally exposed Ser-X-Glu507 motif recognized by the secretory kinase Fam20C. Co-expression of the phosphomimetic PC7-S505E with apoA-V resulted in lower degradation compared to WT, suggesting that Ser505 phosphorylation of PC7 lowers TG levels via reduced apoA-V degradation. In agreement, in Pcsk7-/- mice fed high-fat diet, plasma apoA-V levels and adipocyte LpL activity are increased, providing an in vivo mechanistic link for a role of liver PC7 in enhanced TG storage in adipocytes., (© 2020 Federation of European Biochemical Societies.)- Published
- 2020
- Full Text
- View/download PDF
32. Angiopoietin1 Deficiency in Hepatocytes Affects the Growth of Colorectal Cancer Liver Metastases (CRCLM).
- Author
-
Ibrahim NS, Lazaris A, Rada M, Petrillo SK, Huck L, Hussain S, Ouladan S, Gao ZH, Gregorieff A, Essalmani R, Seidah NG, and Metrakos P
- Abstract
Colorectal cancer liver metastases (CRCLM) that receive their blood supply via vessel co-option are associated with a poor response to anti-angiogenic therapy. Angiopoietins (Ang1 and Ang2) with their Tyrosine-protein kinase receptor (Tie2) have been shown to support vessel co-option. We demonstrate significantly higher expression of Ang1 in hepatocytes adjacent to the tumor region of human chemonaïve and treated co-opting (replacement histopathological growth patterns: RHGP) tumors. To investigate the role of the host Ang1 expression, Ang1 knockout (KO) mice were injected intra-splenically with metastatic MC-38 colon cancer cells that develop co-opting liver metastases. We observed a reduction in the number of liver metastases and interestingly, for the first time, the development of angiogenic driven desmoplastic (DHGP) liver metastases. In addition, in-vitro, knockout of Ang1 in primary hepatocytes inhibited viability, migration and invasion ability of MC-38 cells. We also demonstrate that Ang 1 alone promotes the migration and growth of both human and mouse colon cancer cell lines These results provide evidence that high expression of Ang1 in the host liver is important to support vessel co-option (RHGP lesions) and when inhibited, favours the formation of angiogenic driven liver metastases (DHGP lesions)., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
33. Ser-Phosphorylation of PCSK9 (Proprotein Convertase Subtilisin-Kexin 9) by Fam20C (Family With Sequence Similarity 20, Member C) Kinase Enhances Its Ability to Degrade the LDLR (Low-Density Lipoprotein Receptor).
- Author
-
Ben Djoudi Ouadda A, Gauthier MS, Susan-Resiga D, Girard E, Essalmani R, Black M, Marcinkiewicz J, Forget D, Hamelin J, Evagelidis A, Ly K, Day R, Galarneau L, Corbin F, Coulombe B, Çaku A, Tagliabracci VS, and Seidah NG
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Hep G2 Cells, Hepatocytes metabolism, Humans, Hyperlipoproteinemia Type II physiopathology, In Situ Hybridization methods, Male, Mice, Mice, Knockout, Microscopy, Confocal, Phosphorylation genetics, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction methods, Receptors, LDL metabolism, Sensitivity and Specificity, Calcium-Binding Proteins genetics, Extracellular Matrix Proteins genetics, Gene Expression Regulation, Hyperlipoproteinemia Type II genetics, Proprotein Convertase 9 metabolism, Receptors, LDL genetics
- Abstract
Objective: PCSK9 (proprotein convertase subtilisin-kexin 9) enhances the degradation of the LDLR (low-density lipoprotein receptor) in endosomes/lysosomes. This study aimed to determine the sites of PCSK9 phosphorylation at Ser-residues and the consequences of such posttranslational modification on the secretion and activity of PCSK9 on the LDLR. Approach and Results: Fam20C (family with sequence similarity 20, member C) phosphorylates serines in secretory proteins containing the motif S-X-E/phospho-Ser, including the cholesterol-regulating PCSK9. In situ hybridization of Fam20C mRNA during development and in adult mice revealed a wide tissue distribution, including liver, but not small intestine. Here, we show that Fam20C phosphorylates PCSK9 at Serines 47, 666, 668, and 688. In hepatocytes, phosphorylation enhances PCSK9 secretion and maximizes its induced degradation of the LDLR via the extracellular and intracellular pathways. Replacing any of the 4 Ser by the phosphomimetic Glu or Asp enhanced PCSK9 activity only when the other sites are phosphorylated, whereas Ala substitutions reduced it, as evidenced by Western blotting, Elisa, and LDLR-immunolabeling. This newly uncovered PCSK9/LDLR regulation mechanism refines our understanding of the implication of global PCSK9 phosphorylation in the modulation of LDL-cholesterol and rationalizes the consequence of natural mutations, for example, S668R and E670G. Finally, the relationship of Ser-phosphorylation to the implication of PCSK9 in regulating LDL-cholesterol in the neurological Fragile X-syndrome disorder was investigated., Conclusions: Ser-phosphorylation of PCSK9 maximizes both its secretion and activity on the LDLR. Mass spectrometric approaches to measure such modifications were developed and applied to quantify the levels of bioactive PCSK9 in human plasma under normal and pathological conditions.
- Published
- 2019
- Full Text
- View/download PDF
34. A single domain antibody against the Cys- and His-rich domain of PCSK9 and evolocumab exhibit different inhibition mechanisms in humanized PCSK9 mice.
- Author
-
Essalmani R, Weider E, Marcinkiewicz J, Chamberland A, Susan-Resiga D, Roubtsova A, Seidah NG, and Prat A
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Cysteine metabolism, Genotype, Histidine metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Proprotein Convertase 9 deficiency, Proprotein Convertase 9 metabolism, Antibodies, Monoclonal pharmacology, Cysteine antagonists & inhibitors, Histidine antagonists & inhibitors, PCSK9 Inhibitors
- Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted protein that binds and escorts the low density lipoprotein receptor (LDLR) into the lysosomal degradation pathway. Prescribed monoclonal antibodies (mAbs) against PCSK9 prevent its binding to the LDLR, and result in ~60% lower LDL cholesterol (LDLc) levels. Although efficient, mAbs are expensive. Hence other PCSK9 inhibitors are needed. For screening purpose, we developed C57BL/6J mice expressing the human PCSK9 gene under the control of its own promoter, but lacking endogenous mouse PCSK9. All lines recapitulate the endogenous PCSK9 expression pattern. The Tg2 line that expresses physiological levels of human PCSK9 (hPCSK9) was selected to characterize the inhibitory properties of a previously reported single domain antibody (sdAb), PKF8-mFc, which binds the C-terminal domain of PCSK9. Upon intraveinous injection of 10 mg/kg, PKF8-mFc and the mAb evolocumab neutralized ~50% and 100% of the hPCSK9 impact on total cholesterol (TC) levels, respectively, but PKF8-mFc had a more sustained effect. PKF8-mFc barely affected hPCSK9 levels, whereas evolocumab promoted a 4-fold increase 3 days post-injection, suggesting very different inhibitory mechanisms. The present study also shows that the new transgenic mice are well suited to screen a variety of hPCSK9 inhibitors.
- Published
- 2018
- Full Text
- View/download PDF
35. Osteopontin as a novel substrate for the proprotein convertase 5/6 (PCSK5) in bone.
- Author
-
Hoac B, Susan-Resiga D, Essalmani R, Marcinkiweicz E, Seidah NG, and McKee MD
- Subjects
- Animals, Calcification, Physiologic physiology, Humans, Mice, Mice, Knockout, Substrate Specificity, Bone and Bones metabolism, Osteopontin metabolism, Proprotein Convertase 5 metabolism
- Abstract
Seven proprotein convertases cleave the basic amino acid consensus sequence K/R-X
n -K/R↓ (where n=0, 2, 4 or 6 variable amino acids) to activate precursor proteins. Despite similarities in substrate specificity, basic amino acid-specific proprotein convertases have a distinct tissue distribution allowing for enzymatic actions on tissue-resident substrates. Proprotein convertase 5/6 (PC5/6) has two splice variants - soluble PC5/6A and membrane-bound PC5/6B - and is expressed during mouse development in many tissues including bone and tooth, but little is known about the substrates for PC5/6 therein. Osteopontin (OPN) is an abundant bone extracellular matrix protein with roles in mineralization, cell adhesion and cell migration, and it has putative consensus sequence sites for cleavage by PC5/6, which may modify its function in bone. Since PC5/6-knockout mouse embryos show developmental abnormalities, and reduced overall mineralization, we designed this study to determine whether OPN is a substrate of PC5/6. In silico analysis of OPN protein sequences identified four potential PC5/6 consensus cleavage sites in human OPN, and three sites - including a noncanonical sequence - in mouse OPN. Ex vivo co-transfections with human OPN revealed complete OPN cleavage reducing full-length OPN (~70kDa) to an N-terminal fragment migrating at ~50kDa and two C-terminal fragments at ~18kDa and ~16kDa. Direct cleavage of OPN by PC5/6A - the predominant isoform expressed in human osteoblast cells - was confirmed by cell-free enzyme-substrate assays and by mass spectrometry. The latter was also used to investigate potential cleavage sites. Co-transfections of PC5/6 and mouse OPN showed partial cleavage of OPN into a C-terminal OPN fragment migrating at ~30kDa and an N-terminal fragment migrating at ~29kDa. Micro-computed tomography of PC5/6-knockout embryos at E18.5 confirmed a reduction in mineralized bone, and in situ hybridization performed on cryo-sections of normal mouse bone using Pcsk5 and Opn anti-sense and control-sense cRNA probes indicated the co-localization of the expression of these genes in bone cells. This mRNA expression profile was supported by semi-quantitative RT-PCR using osteoblast primary cultures, and cultured MC3T3-E1 osteoblast and MLO-Y4 osteocyte cell lines. Immunoblotting for OPN from mouse bone extracts showed altered OPN processing in PC5/6-knockout mice compared to wildtype mice. OPN fragments migrated at ~25kDa and ~16kDa in wildtype bone and were not present in PC5/6-deficient bone. In conclusion, this study demonstrates that Pcsk5 is expressed in bone-forming cells, and that OPN is a novel substrate for PC5/6. Cleavage of OPN by PC5/6 may modify the function of OPN in bone and/or modulate other enzymatic cleavages of OPN, leading to alterations in the bone phenotype., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
36. Proprotein convertase furin regulates osteocalcin and bone endocrine function.
- Author
-
Al Rifai O, Chow J, Lacombe J, Julien C, Faubert D, Susan-Resiga D, Essalmani R, Creemers JW, Seidah NG, and Ferron M
- Subjects
- Amino Acid Sequence, Animals, Bone and Bones cytology, Cells, Cultured, Endocrine System, Energy Metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Osteoblasts enzymology, Proprotein Convertase 5 metabolism, Protein Processing, Post-Translational, Protein Transport, Proteolysis, RAW 264.7 Cells, Bone and Bones enzymology, Furin physiology, Osteocalcin metabolism
- Abstract
Osteocalcin (OCN) is an osteoblast-derived hormone that increases energy expenditure, insulin sensitivity, insulin secretion, and glucose tolerance. The cDNA sequence of OCN predicts that, like many other peptide hormones, OCN is first synthesized as a prohormone (pro-OCN). The importance of pro-OCN maturation in regulating OCN and the identity of the endopeptidase responsible for pro-OCN cleavage in osteoblasts are still unknown. Here, we show that the proprotein convertase furin is responsible for pro-OCN maturation in vitro and in vivo. Using pharmacological and genetic experiments, we also determined that furin-mediated pro-OCN cleavage occurred independently of its γ-carboxylation, a posttranslational modification that is known to hamper OCN endocrine action. However, because pro-OCN is not efficiently decarboxylated and activated during bone resorption, inactivation of furin in osteoblasts in mice resulted in decreased circulating levels of undercarboxylated OCN, impaired glucose tolerance, and reduced energy expenditure. Furthermore, we show that Furin deletion in osteoblasts reduced appetite, a function not modulated by OCN, thus suggesting that osteoblasts may secrete additional hormones that regulate different aspects of energy metabolism. Accordingly, the metabolic defects of the mice lacking furin in osteoblasts became more apparent under pair-feeding conditions. These findings identify furin as an important regulator of bone endocrine function.
- Published
- 2017
- Full Text
- View/download PDF
37. Thrombin activation of protein C requires prior processing by a liver proprotein convertase.
- Author
-
Essalmani R, Susan-Resiga D, Guillemot J, Kim W, Sachan V, Awan Z, Chamberland A, Asselin MC, Ly K, Desjardins R, Day R, Prat A, and Seidah NG
- Published
- 2017
- Full Text
- View/download PDF
38. The Proprotein Convertase Subtilisin/Kexin Type 9-resistant R410S Low Density Lipoprotein Receptor Mutation: A NOVEL MECHANISM CAUSING FAMILIAL HYPERCHOLESTEROLEMIA.
- Author
-
Susan-Resiga D, Girard E, Kiss RS, Essalmani R, Hamelin J, Asselin MC, Awan Z, Butkinaree C, Fleury A, Soldera A, Dory YL, Baass A, and Seidah NG
- Subjects
- Amino Acid Substitution, Endosomes genetics, Female, Humans, Lysosomes genetics, Male, Mutation, Missense, Protein Binding, Endosomes metabolism, Hyperlipoproteinemia Type II genetics, Hyperlipoproteinemia Type II metabolism, Lipoproteins, LDL metabolism, Lysosomes metabolism, Proprotein Convertase 9 metabolism, Receptors, LDL genetics, Receptors, LDL metabolism
- Abstract
Familial hypercholesterolemia (FH) is characterized by severely elevated low density lipoprotein (LDL) cholesterol. Herein, we identified an FH patient presenting novel compound heterozygote mutations R410S and G592E of the LDL receptor (LDLR). The patient responded modestly to maximum rosuvastatin plus ezetimibe therapy, even in combination with a PCSK9 monoclonal antibody injection. Using cell biology and molecular dynamics simulations, we aimed to define the underlying mechanism(s) by which these LDLR mutations affect LDL metabolism and lead to hypercholesterolemia. Our data showed that the LDLR-G592E is a class 2b mutant, because it mostly failed to exit the endoplasmic reticulum and was degraded. Even though LDLR-R410S and LDLR-WT were similar in levels of cell surface and total receptor and bound equally well to LDL or extracellular PCSK9, the LDLR-R410S was resistant to exogenous PCSK9-mediated degradation in endosomes/lysosomes and showed reduced LDL internalization and degradation relative to LDLR-WT. Evidence is provided for a tighter association of LDL with LDLR-R410S at acidic pH, a reduced LDL delivery to late endosomes/lysosomes, and an increased release in the medium of the bound/internalized LDL, as compared with LDLR-WT. These data suggested that LDLR-R410S recycles loaded with its LDL-cargo. Our findings demonstrate that LDLR-R410S represents an LDLR loss-of-function through a novel class 8 FH-causing mechanism, thereby rationalizing the observed phenotype., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
39. Proprotein convertase subtilisin/Kexin type 9 (PCSK9) single domain antibodies are potent inhibitors of low density lipoprotein receptor degradation.
- Author
-
Weider E, Susan-Resiga D, Essalmani R, Hamelin J, Asselin MC, Nimesh S, Ashraf Y, Wycoff KL, Zhang J, Prat A, and Seidah NG
- Published
- 2016
- Full Text
- View/download PDF
40. An Unbiased Mass Spectrometry Approach Identifies Glypican-3 as an Interactor of Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) and Low Density Lipoprotein Receptor (LDLR) in Hepatocellular Carcinoma Cells.
- Author
-
Ly K, Essalmani R, Desjardins R, Seidah NG, and Day R
- Subjects
- Carcinoma, Hepatocellular genetics, Glypicans genetics, Hep G2 Cells, Humans, Lipoproteins, LDL genetics, Lipoproteins, LDL metabolism, Liver Neoplasms genetics, Matrilin Proteins genetics, Matrilin Proteins metabolism, Neoplasm Proteins genetics, Proprotein Convertase 9 genetics, Protein Binding, Receptors, LDL genetics, Carcinoma, Hepatocellular metabolism, Glypicans metabolism, Liver Neoplasms metabolism, Neoplasm Proteins metabolism, Proprotein Convertase 9 metabolism, Receptors, LDL metabolism
- Abstract
The mechanism of LDL receptor (LDLR) degradation mediated by the proprotein convertase subtilisin/kexin type 9 (PCSK9) has been extensively studied; however, many steps within this process remain unclear and still require characterization. Recent studies have shown that PCSK9 lacking its Cys/His-rich domain can still promote LDLR internalization, but the complex does not reach the lysosome suggesting the presence of an additional interaction partner(s). In this study we carried out an unbiased screening approach to identify PCSK9-interacting proteins in the HepG2 cells' secretome using co-immunoprecipitation combined with mass spectrometry analyses. Several interacting proteins were identified, including glypican-3 (GPC3), phospholipid transfer protein, matrilin-3, tissue factor pathway inhibitor, fibrinogen-like 1, and plasminogen activator inhibitor-1. We then validated these interactions by co-immunoprecipitation and Western blotting. Furthermore, functional validation was examined by silencing each candidate protein in HepG2 cells using short hairpin RNAs to determine their effect on LDL uptake and LDLR levels. Only GPC3 and phospholipid transfer protein silencing in HepG2 cells significantly increased LDL uptake in these cells and displayed higher total LDLR protein levels compared with control cells. Moreover, our study provides the first evidence that GPC3 can modulate the PCSK9 extracellular activity as a competitive binding partner to the LDLR in HepG2 cells., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
41. Deferoxamine stimulates LDLR expression and LDL uptake in HepG2 cells.
- Author
-
Guillemot J, Asselin MC, Susan-Resiga D, Essalmani R, and Seidah NG
- Subjects
- Cholesterol metabolism, Hep G2 Cells drug effects, Hep G2 Cells metabolism, Humans, Hydroxymethylglutaryl CoA Reductases genetics, Hydroxymethylglutaryl CoA Reductases metabolism, Iron metabolism, Iron Chelating Agents pharmacology, Lipoproteins, LDL pharmacokinetics, Proprotein Convertase 9 genetics, Proprotein Convertase 9 metabolism, RNA Processing, Post-Transcriptional, Receptors, LDL metabolism, Sterol Regulatory Element Binding Protein 1 genetics, Sterol Regulatory Element Binding Protein 1 metabolism, Sterol Regulatory Element Binding Protein 2 genetics, Sterol Regulatory Element Binding Protein 2 metabolism, Deferoxamine pharmacology, Lipoproteins, LDL metabolism, Receptors, LDL genetics
- Abstract
Scope: Iron overload contributes to the pathogenesis of atherosclerosis and iron chelators are beneficial through their antioxidant properties. Hepatic iron loading increases cholesterol synthesis. Whether iron depletion could affect hepatic cholesterol metabolism is unknown., Methods and Results: We examined the effect of the iron chelator deferoxamine (DFO) on mRNA expression of genes involved in cholesterol metabolism and/or cholesterol uptake. Our results revealed that DFO increases LDL receptor (LDLR) mRNA levels in human hepatocyte-derived cell lines HepG2 and Huh7 cells, and in K562 cells. In HepG2 cells, we observed that DFO increases (i) LDLR-mRNA levels in a time- and dose-dependent manner, (ii) LDLR-protein levels; (iii) cell surface LDLR; and (iv) LDL uptake. In contrast, the mRNA levels of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, sterol regulatory element-binding proteins, and the mRNA/protein levels of proprotein convertase subtilisin-kexin 9 were not modulated by DFO, suggesting that the LDLR regulation by DFO is not at the transcriptional or posttranslational levels. Since LDLR-mRNA was stabilized by DFO, a posttranscriptional mechanism is suggested for the DFO-mediated upregulation of LDLR., Conclusion: DFO induced an increase in LDLR expression by a posttranscriptional mechanism resulting in an enhancement of LDL uptake in HepG2 cells, suggesting increased LDLR activity as one of the underlying causes of the hypocholesterolemic effect of iron reduction., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2016
- Full Text
- View/download PDF
42. Reducing Vascular Calcification by Anti-IL-1β Monoclonal Antibody in a Mouse Model of Familial Hypercholesterolemia.
- Author
-
Awan Z, Denis M, Roubtsova A, Essalmani R, Marcinkiewicz J, Awan A, Gram H, Seidah NG, and Genest J
- Subjects
- Animals, Aorta diagnostic imaging, Aorta metabolism, Aorta physiopathology, Aortic Diseases blood, Aortic Diseases diagnosis, Aortic Diseases genetics, Aortic Diseases immunology, Aortic Diseases physiopathology, Aortography methods, Biomarkers blood, Blood Flow Velocity, Disease Models, Animal, Genetic Predisposition to Disease, Hyperlipoproteinemia Type II blood, Hyperlipoproteinemia Type II genetics, Hyperlipoproteinemia Type II immunology, Interleukin-1beta blood, Interleukin-1beta immunology, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Proprotein Convertases genetics, Proprotein Convertases metabolism, Receptors, LDL deficiency, Receptors, LDL genetics, Regional Blood Flow, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, Ultrasonography, Vascular Calcification diagnosis, Vascular Calcification genetics, Vascular Calcification immunology, Vascular Calcification metabolism, Vascular Calcification physiopathology, X-Ray Microtomography, Anti-Inflammatory Agents pharmacology, Antibodies, Monoclonal pharmacology, Aorta drug effects, Aortic Diseases prevention & control, Hyperlipoproteinemia Type II drug therapy, Interleukin-1beta antagonists & inhibitors, Vascular Calcification prevention & control
- Abstract
Background: Given the link between cholesterol and activation of inflammation via interleukin 1β (IL-1β), we tested the effects of IL-1β inhibition on atherosclerotic calcification in mice. Patients with familial hypercholesterolemia develop extensive aortic calcification and calcific aortic stenosis. Although statins delay this process, low-density lipoprotein (LDL) cholesterol lowering alone is not enough to avert it. Data suggest that vascular inflammation initiated by hypercholesterolemia is followed by unchecked mineralization at sites of atherosclerotic plaques. The LDL-receptor (LDLR)-deficient (Ldlr(-/-)) and LDLR-attenuated Pcsk9(Tg) mice are available animal models for pharmacological testing., Methods: A mouse monoclonal antibody (mAb) against IL-1β or placebo was administered subcutaneously in Ldlr(-/-) and Pcsk9(Tg) models fed a Western diet. Drug level, anthropometric, lipid, and glucose profiles were determined. Expressions of proprotein convertase subtilisin/kexin type 9 (PCSK9), serum amyloid A1, and cytokine were measured by enzyme-linked immunosorbent assay. Aortic calcification was determined by microcomputerized tomography (micro-CT) and X-ray densitometry, and aortic flow velocity was assessed by ultrasound., Results: Circulating levels of IL-1β in Ldlr(-/-) mice were significantly greater (2-fold) than observed in Pcsk9(Tg) mice. Placebo- and mAb-treated mice did not differ in their growth, lipid, glucose profiles, and other cytokines. Calcifications were significantly diminished in mAb-treatment Ldlr(-/-) mice (a reduction of ∼ 75% by X-ray and ∼ 90% by micro-CT) and reduced insignificantly in mAb-treatment Pcsk9(Tg) mice, whereas aortic flow velocity was unchanged in both models., Conclusions: Herein, we demonstrate that aortic calcifications can be inhibited by an IL-1β mAb in LDLR-deficient mice. These results have a translational component to prevent vascular calcification in human and represent new evidence to rationalize targeting inflammation in cardiovascular disease., (© The Author(s) 2015.)
- Published
- 2016
- Full Text
- View/download PDF
43. PCSK9 deficiency unmasks a sex- and tissue-specific subcellular distribution of the LDL and VLDL receptors in mice.
- Author
-
Roubtsova A, Chamberland A, Marcinkiewicz J, Essalmani R, Fazel A, Bergeron JJ, Seidah NG, and Prat A
- Subjects
- Adiposity, Animals, Estradiol physiology, Female, Intra-Abdominal Fat metabolism, Liver enzymology, Male, Mice, Inbred C57BL, Mice, Knockout, Organ Specificity, Proprotein Convertase 9, Proprotein Convertases blood, Proprotein Convertases deficiency, Protein Transport, RNA, Messenger genetics, RNA, Messenger metabolism, Serine Endopeptidases blood, Serine Endopeptidases deficiency, Sex Characteristics, Proprotein Convertases genetics, Receptors, LDL metabolism, Serine Endopeptidases genetics
- Abstract
Proprotein convertase subtilisin kexin type 9 (PCSK9), the last member of the family of Proprotein Convertases related to Subtilisin and Kexin, regulates LDL-cholesterol by promoting the endosomal/lysosomal degradation of the LDL receptor (LDLR). Herein, we show that the LDLR cell surface levels dramatically increase in the liver and pancreatic islets of PCSK9 KO male but not female mice. In contrast, in KO female mice, the LDLR is more abundant at the cell surface enterocytes, as is the VLDL receptor (VLDLR) at the cell surface of adipocytes. Ovariectomy of KO female mice led to a typical KO male pattern, whereas 17β-estradiol (E2) treatment restored the female pattern without concomitant changes in LDLR adaptor protein 1 (also known as ARH), disabled-2, or inducible degrader of the LDLR expression levels. We also show that this E2-mediated regulation, which is observed only in the absence of PCSK9, is abolished upon feeding the mice a high-cholesterol diet. The latter dramatically represses PCSK9 expression and leads to high surface levels of the LDLR in the hepatocytes of all sexes and genotypes. In conclusion, the absence of PCSK9 results in a sex- and tissue-specific subcellular distribution of the LDLR and VLDLR, which is determined by E2 levels., (Copyright © 2015 by the American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
44. Neuroinflammation-Induced Interactions between Protease-Activated Receptor 1 and Proprotein Convertases in HIV-Associated Neurocognitive Disorder.
- Author
-
Kim W, Zekas E, Lodge R, Susan-Resiga D, Marcinkiewicz E, Essalmani R, Mihara K, Ramachandran R, Asahchop E, Gelman B, Cohen ÉA, Power C, Hollenberg MD, and Seidah NG
- Subjects
- Amino Acid Sequence, Animals, Brain metabolism, Cell Line, Furin genetics, Gene Expression Regulation, HIV Envelope Protein gp160 metabolism, HIV Infections genetics, HIV Infections metabolism, HIV Infections pathology, HIV-1 physiology, Host-Pathogen Interactions, Humans, Inflammation genetics, Inflammation metabolism, Inflammation pathology, Mice, Molecular Sequence Data, Neurocognitive Disorders genetics, Neurocognitive Disorders metabolism, Neurocognitive Disorders pathology, Proprotein Convertase 5 analysis, Proprotein Convertase 5 metabolism, Proprotein Convertases analysis, Proprotein Convertases genetics, RNA, Messenger analysis, RNA, Messenger genetics, Receptor, PAR-1 analysis, Receptor, PAR-1 genetics, Serine Endopeptidases analysis, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, Subtilisins analysis, Subtilisins genetics, Subtilisins metabolism, Thrombin metabolism, Brain pathology, HIV Infections complications, Inflammation complications, Neurocognitive Disorders complications, Proprotein Convertases metabolism, Protein Interaction Maps, Receptor, PAR-1 metabolism
- Abstract
The proprotein convertases (PCs) furin, PC5, PACE4, and PC7 cleave secretory proteins after basic residues, including the HIV envelope glycoprotein (gp160) and Vpr. We evaluated the abundance of PC mRNAs in postmortem brains of individuals exhibiting HIV-associated neurocognitive disorder (HAND), likely driven by neuroinflammation and neurotoxic HIV proteins (e.g., envelope and Vpr). Concomitant with increased inflammation-related gene expression (interleukin-1β [IL-1β]), the mRNA levels of the above PCs are significantly increased, together with those of the proteinase-activated receptor 1 (PAR1), an inflammation-associated receptor that is cleaved by thrombin at ProArg41↓ (where the down arrow indicates the cleavage location), and potentially by PCs at Arg41XXXXArg46↓. The latter motif in PAR1, but not its R46A mutant, drives its interactions with PCs. Indeed, PAR1 upregulation leads to the inhibition of membrane-bound furin, PC5B, and PC7 and inhibits gp160 processing and HIV infectivity. Additionally, a proximity ligation assay revealed that furin and PC7 interact with PAR1. Reciprocally, increased furin expression reduces the plasma membrane abundance of PAR1 by trapping it in the trans-Golgi network. Furthermore, soluble PC5A/PACE4 can target/disarm cell surface PAR1 through cleavage at Arg46↓. PACE4/PC5A decreased calcium mobilization induced by thrombin stimulation. Our data reveal a new PC-PAR1-interaction pathway, which offsets the effects of HIV-induced neuroinflammation, viral infection, and potentially the development of HAND., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
45. Amyloid Precursor-like Protein 2 and Sortilin Do Not Regulate the PCSK9 Convertase-mediated Low Density Lipoprotein Receptor Degradation but Interact with Each Other.
- Author
-
Butkinaree C, Canuel M, Essalmani R, Poirier S, Benjannet S, Asselin MC, Roubtsova A, Hamelin J, Marcinkiewicz J, Chamberland A, Guillemot J, Mayer G, Sisodia SS, Jacob Y, Prat A, and Seidah NG
- Subjects
- Adaptor Proteins, Vesicular Transport biosynthesis, Adaptor Proteins, Vesicular Transport genetics, Amyloid beta-Protein Precursor genetics, Animals, Gene Expression Regulation, Hep G2 Cells, Hepatocytes metabolism, Humans, Liver metabolism, Mice, Nerve Tissue Proteins genetics, Proprotein Convertase 9, Proprotein Convertases genetics, Receptors, LDL genetics, Serine Endopeptidases genetics, Adaptor Proteins, Vesicular Transport metabolism, Amyloid beta-Protein Precursor metabolism, Nerve Tissue Proteins metabolism, Proprotein Convertases metabolism, Proteolysis, Receptors, LDL biosynthesis, Serine Endopeptidases metabolism
- Abstract
Amyloid precursor-like protein 2 (APLP2) and sortilin were reported to individually bind the proprotein convertase subtilisin/kexin type 9 (PCSK9) and regulate its activity on the low-density lipoprotein receptor (LDLR). The data presented herein demonstrate that mRNA knockdowns of APLP2, sortilin, or both in the human hepatocyte cell lines HepG2 and Huh7 do not affect the ability of extracellular PCSK9 to enhance the degradation of the LDLR. Furthermore, mice deficient in APLP2 or sortilin do not exhibit significant changes in liver LDLR or plasma total cholesterol levels. Moreover, cellular overexpression of one or both proteins does not alter PCSK9 secretion, or its activity on the LDLR. We conclude that PCSK9 enhances the degradation of the LDLR independently of either APLP2 or sortilin both ex vivo and in mice. Interestingly, when co-expressed with PCSK9, both APLP2 and sortilin were targeted for lysosomal degradation. Using chemiluminescence proximity and co-immunoprecipitation assays, as well as biosynthetic analysis, we discovered that sortilin binds and stabilizes APLP2, and hence could regulate its intracellular functions on other targets., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
46. Is there a link between proprotein convertase PC7 activity and human lipid homeostasis?
- Author
-
Guillemot J, Essalmani R, Hamelin J, and Seidah NG
- Abstract
A genome-wide association study suggested that a R504H mutation in the proprotein convertase PC7 is associated with increased circulating levels of HDL and reduced triglycerides in black Africans. Our present results show that PC7 and PC7-R504H exhibit similar processing of transferrin receptor-1, proSortilin, and apolipoprotein-F. Plasma analyses revealed no change in the lipid profiles, insulin or glucose of wild type and PC7 KO mice. Thus, the R504H mutation does not modify the proteolytic activity of PC7. The mechanisms behind the implication of PC7 in the regulation of human HDL, triglycerides and in modifying the levels of atherogenic small dense LDL remain to be elucidated.
- Published
- 2014
- Full Text
- View/download PDF
47. Disruption of the expression of the proprotein convertase PC7 reduces BDNF production and affects learning and memory in mice.
- Author
-
Wetsel WC, Rodriguiz RM, Guillemot J, Rousselet E, Essalmani R, Kim IH, Bryant JC, Marcinkiewicz J, Desjardins R, Day R, Constam DB, Prat A, and Seidah NG
- Subjects
- Amygdala metabolism, Animals, Blotting, Western, COS Cells, Cells, Cultured, Chlorocebus aethiops, Female, Flavanones pharmacology, Gene Expression, HEK293 Cells, Hepatocytes cytology, Hepatocytes metabolism, Hippocampus metabolism, Humans, In Situ Hybridization, Male, Maze Learning drug effects, Memory drug effects, Mice, Mice, Knockout, Protein Precursors genetics, Protein Precursors metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Subtilisins genetics, Brain-Derived Neurotrophic Factor metabolism, Maze Learning physiology, Memory physiology, Subtilisins metabolism
- Abstract
PC7 belongs to the proprotein convertase family, whose members are implicated in the cleavage of secretory precursors. The in vivo function of PC7 is unknown. Herein, we find that the precursor proBDNF is processed into mature BDNF in COS-1 cells coexpressing proBDNF with either PC7 or Furin. Conversely, the processing of proBDNF into BDNF is markedly reduced in the absence of either Furin or PC7 in mouse primary hepatocytes. In vivo we observe that BDNF and PC7 mRNAs are colocalized in mouse hippocampus and amygdala and that mature BDNF protein levels are reduced in these brain areas in PC7 KO mice but not in the hippocampus of PC1/3 KO mice. Various behavioral tests reveal that in PC7 KO mice spatial memory is intact and plasticity of responding is mildly abnormal. Episodic and emotional memories are severely impaired, but both are rescued with the tyrosine receptor kinase B agonist 7,8-dihydroxyflavone. Altogether, these results support an in vivo role for PC7 in the regulation of certain types of cognitive performance, in part via proBDNF processing. Because polymorphic variants of human PC7 are being characterized, it will be important in future studies to determine their effects on additional physiological and behavioral processes.
- Published
- 2013
- Full Text
- View/download PDF
48. Processing of human toll-like receptor 7 by furin-like proprotein convertases is required for its accumulation and activity in endosomes.
- Author
-
Hipp MM, Shepherd D, Gileadi U, Aichinger MC, Kessler BM, Edelmann MJ, Essalmani R, Seidah NG, Reis e Sousa C, and Cerundolo V
- Subjects
- Amino Acid Sequence, Autoimmunity, Cell Line, Endosomes drug effects, Endosomes immunology, Feedback, Physiological, Furin genetics, Furin immunology, Gene Expression Regulation, Genetic Vectors, Humans, Lentivirus genetics, Macrophages cytology, Macrophages drug effects, Macrophages immunology, Molecular Sequence Data, Orthomyxoviridae immunology, Proprotein Convertases genetics, Proprotein Convertases immunology, Protein Structure, Tertiary, Quinolines pharmacology, Signal Transduction, Toll-Like Receptor 7 genetics, Toll-Like Receptor 7 immunology, Furin metabolism, Macrophages metabolism, Proprotein Convertases metabolism, Protein Processing, Post-Translational, Toll-Like Receptor 7 metabolism
- Abstract
Toll-like receptor 7 (TLR7) triggers antiviral immune responses by recognizing viral single-stranded RNA in endosomes, but the biosynthetic pathway of human TLR7 (hTLR7) remains unclear. Here, we show that hTLR7 is proteolytically processed and that the C-terminal fragment selectively accumulates in endocytic compartments. hTLR7 processing occurred at neutral pH and was dependent on furin-like proprotein convertases (PCs). Furthermore, TLR7 processing was required for its functional response to TLR7 agonists such as R837 or influenza virus. Notably, proinflammatory and differentiation stimuli increased the expression of furin-like PCs in immune cells, suggesting a positive feedback mechanism for TLR7 processing during infection. Because self-RNA can under certain conditions activate TLR7 and trigger autoimmunity, our results identify furin-like PCs as a possible target to attenuate TLR7-dependent autoimmunity and other immune pathologies., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
49. Furin is the primary in vivo convertase of angiopoietin-like 3 and endothelial lipase in hepatocytes.
- Author
-
Essalmani R, Susan-Resiga D, Chamberland A, Asselin MC, Canuel M, Constam D, Creemers JW, Day R, Gauthier D, Prat A, and Seidah NG
- Subjects
- Angiopoietin-Like Protein 3, Angiopoietin-like Proteins, Animals, COS Cells, Chlorocebus aethiops, Cholesterol, HDL metabolism, Furin genetics, Gene Silencing, Lipoproteins, HDL metabolism, Male, Mice, Mice, Knockout, Phospholipids metabolism, Proprotein Convertase 5 genetics, Angiopoietins metabolism, Furin metabolism, Hepatocytes enzymology, Lipase metabolism
- Abstract
The proprotein convertases (PCs) furin, PC5/6, and PACE4 exhibit unique and/or complementary functions. Their knock-out (KO) in mice resulted in strong and specific phenotypes demonstrating that, in vivo, these PCs are unique and essential during development. However, they also exhibit redundant functions. Liver angiopoietin-like 3 (ANGPTL3) inhibits lipolysis by binding to lipoprotein lipases. It is found in the plasma as full length and truncated forms. The latter is more active and generated by cleavage at a furin-like site. Endothelial lipase (EL) binds heparin sulfate proteoglycans on cell surfaces and catalyzes the hydrolysis of HDL phospholipids. EL activity is regulated by two endogenous inhibitors, ANGPTL3 and ANGPTL4, and by PCs that inactivate EL through cleavage releasing the N-terminal catalytic and C-terminal lipid-binding domains. Herein, because furin and PC5/6 complete KOs are lethal, we used mice lacking furin or PC5/6 specifically in hepatocytes (hKO) or mice completely lacking PACE4. In primary hepatocytes, ANGPTL3 was processed into a shorter form of ANGPTL3 intracellularly by furin only, and extracellularly mainly by PACE4. In vivo, the absence of furin in hepatocytes reduced by ∼50% the circulating levels of cleaved ANGPTL3, while the lack of PACE4 had only a minor effect. Analysis of the EL processing in primary hepatocytes and in vivo revealed that it is mostly cleaved by furin. However, the lack of furin or PC5/6 in hepatocytes and complete PACE4 KO did not appreciably modify plasma HDL levels or EL activity. Thus, inhibition of furin in liver would not be expected to modify the plasma lipid profiles.
- Published
- 2013
- Full Text
- View/download PDF
50. Implication of the proprotein convertases in iron homeostasis: proprotein convertase 7 sheds human transferrin receptor 1 and furin activates hepcidin.
- Author
-
Guillemot J, Canuel M, Essalmani R, Prat A, and Seidah NG
- Subjects
- Animals, COS Cells, Chlorocebus aethiops, Down-Regulation, Endosomes metabolism, HEK293 Cells, Hep G2 Cells, Hepcidins, Humans, Mice, Protein Structure, Tertiary, Antigens, CD metabolism, Antimicrobial Cationic Peptides metabolism, Furin metabolism, Hepatocytes metabolism, Iron metabolism, Receptors, Transferrin metabolism, Subtilisins metabolism
- Abstract
Unlabelled: The first seven members of the proprotein convertase (PC) family activate protein precursors by cleavage after basic residues. While PC7 has no known specific substrates, it shows redundancy with other PCs. A genome-wide association study suggested that circulating levels of shed human transferrin receptor 1 (hTfR1) are regulated by PC7. We thus examined whether hTfR1 constitutes a specific substrate for PC7. Coexpression of hTfR1 with PCs in several cell lines indicated that PC7 is the only convertase that sheds this receptor into the medium. Site-directed mutagenesis showed that cleavage occurs at the unusual site KTECER100 ↓LA, in which the P1 Arg100 and P6 Lys95 are critical. Pharmacological treatments revealed that shedding of hTfR1 by PC7 requires endocytosis into acidic clathrin-coated vesicles. A PC7 chimera, in which the transmembrane domain and the cytosolic tail of PC7 were replaced by that of the convertase furin, lost its ability to cleave the receptor, demonstrating the importance of these domains in the regulation of PC7 function. Analysis of primary hepatocytes from mice lacking furin, PC5, PACE4, or PC7 revealed that hepcidin, which limits iron availability in the circulation, is specifically generated by furin and not by PC7. Finally, depletion of iron in the medium of hepatoma cell lines incubated with the iron chelator desferrioxamine resulted in PC7 down-regulation., Conclusion: Among the PC family members, only furin activates hepcidin in hepatocytes, and uniquely the full-length membrane-bound PC7 can directly shed hTfR1 by cleavage at Arg100 ↓. Our results support the notion that, when iron is limiting, hTfR1 levels increase at least in part by way of the down-regulation of PC7 expression. (HEPATOLOGY 2013;)., (Copyright © 2013 American Association for the Study of Liver Diseases.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.